Human CD4+ T cells specific for Merkel cell polyomavirus localize to Merkel cell carcinomas and target a required oncogenic domain

immuneACCESS ◽  
2019 ◽  
Author(s):  
NV Longino ◽  
J Yang ◽  
JG Iyer ◽  
D Ibrani ◽  
I Chow ◽  
...  
2019 ◽  
Vol 7 (10) ◽  
pp. 1727-1739 ◽  
Author(s):  
Natalie V. Longino ◽  
Junbao Yang ◽  
Jayasri G. Iyer ◽  
Dafina Ibrani ◽  
I-Ting Chow ◽  
...  

immuneACCESS ◽  
2018 ◽  
Author(s):  
NJ Miller ◽  
CD Church ◽  
L Dong ◽  
D Crispin ◽  
MP Fitzgibbon ◽  
...  

2020 ◽  
Vol 11 ◽  
Author(s):  
Sarah I. Davies ◽  
John Barrett ◽  
Susan Wong ◽  
Mark Jesse Chang ◽  
Pawel J. Muranski ◽  
...  

Virus positive Merkel cell carcinoma (VP-MCC) is an aggressive but immunogenic skin malignancy driven by Merkel cell polyomavirus (MCPyV) T antigen (TAg). Since adoptive T cell transfer (ACT) can be effective against virus-driven malignancies, we set out to develop a methodology for generating MCPyV TAg specific T cells. MCPyV is a common, asymptomatic infection and virus-exposed healthy donors represent a potential source of MCPyV TAg specific T cells for ACT. Virus specific T cells were generated using monocyte-derived dendritic cells (moDCs) pulsed with MCPyV TAg peptide libraries and co-cultured with autologous T cells in supplemented with pro-inflammatory and homeostatic cytokines for 14 days. Specific reactivity was observed predominantly within the CD4+ T cell compartment in the cultures generated from 21/46 random healthy donors. Notably, responses were more often seen in donors aged 50 years and older. TAg specific CD4+ T cells specifically secreted Th1 cytokines and upregulated CD137 upon challenge with MCPyV TAg peptide libraries and autologous transduced antigen presenting cells. Expanded T cells from healthy donors recognized epitopes of both TAg splice variants found in VP-MCC tumors, and minimally expressed exhaustion markers. Our data show that MCPyV specific T cells can be expanded from healthy donors using methods appropriate for the manufacture of clinical grade ACT products.


2017 ◽  
Vol 5 (2) ◽  
pp. 137-147 ◽  
Author(s):  
Natalie J. Miller ◽  
Candice D. Church ◽  
Lichun Dong ◽  
David Crispin ◽  
Matthew P. Fitzgibbon ◽  
...  

2018 ◽  
Vol 24 (15) ◽  
pp. 3644-3655 ◽  
Author(s):  
Ioannis Gavvovidis ◽  
Matthias Leisegang ◽  
Gerald Willimsky ◽  
Natalie Miller ◽  
Paul Nghiem ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A910-A910
Author(s):  
Claire Buchta Rosean ◽  
Claire Buchta Rosean ◽  
Pratima Sinha ◽  
David Koelle ◽  
Paul Nghiem ◽  
...  

BackgroundThe majority of Merkel cell carcinomas (MCC), a rare and highly-aggressive type of neuroendocrine skin cancer, are associated with Merkel cell polyomavirus (MCPyV) infection. MCPyV integrates into the host genome, resulting in expression of a truncated form of the viral large T antigen (LT) in infected cells, and making LT an attractive target for therapeutic cancer vaccines. While induction of tumor-reactive CD8+ T cells is a major goal of cancer therapy, CD4+ T cells provide essential support to CD8+ T cells by promoting their expression of cytotoxic effector molecules and increasing their migratory capacity. Cytokines secreted by CD4+ T cells, such as IFNγ, can also exert desirable effects on the tumor microenvironment. Therefore, we set out to design a cancer vaccine that promotes potent, antigen-specific CD4+ T cell responses to MCPyV-LT.MethodsTo activate antigen-specific CD4+ T cells in vivo, we utilized our nucleic acid platform, UNITE (UNiversal Intracellular Targeted Expression), which fuses a tumor-associated antigen with lysosomal-associated membrane protein 1 (LAMP1). This lysosomal targeting technology results in enhanced antigen presentation and a balanced T cell response. LTS220A, encoding a mutated form of MCPyV-LT that abrogates its pro-oncogenic properties, was introduced into the UNITE platform. LTS220A-UNITE, known as ITI-3000, was administered to female C57BL/6 mice intradermally in the ear with electroporation.ResultsITI-3000 promoted a potent, antigen-specific CD4+ T cell response to MCPyV-LT. Vaccination with ITI-3000 significantly delayed and slowed growth of B16F10 tumors expressing LTS220A in prophylactic and therapeutic settings, respectively. ITI-3000 induced a favorable tumor microenvironment (TME), including significantly enhanced numbers of CD4+ T cells, CD8+ T cells, NK cells, and NKT cells. Tumor-infiltrating myeloid cells were reduced in frequency in vaccinated mice and polarized towards an anti-tumor phenotype. Cytokine analysis of the TME showed significantly enhanced levels of cytokines associated with anti-tumor immune responses in ITI-3000-vaccinated mice, including IFNγ, TNFα, IL-2, and IL-1β. Additionally, ITI-3000 synergized with PD-1 blockade, further reducing tumor burden and enhancing survival in mice receiving combination therapy.ConclusionsWe find that DNA vaccination with ITI-3000 using the UNITE platform enhances CD4+ T cell responses to MCPyV-LT and results in anti-tumor immune responses in a mouse model of Merkel cell carcinoma.Ethics ApprovalThis study was approved by Immunomic Therapeutics’ Institutional Animal Care and Use Committee, protocol number 16-11-002.


Sign in / Sign up

Export Citation Format

Share Document